Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Mol Med Rep ; 26(6)2022 Dec.
Article in English | MEDLINE | ID: mdl-36300202

ABSTRACT

Cathepsins are one of the most abundant proteases within the lysosomes with diverse physiological effects ranging from immune responses, cell death and intracellular protein degradation. Cathepsins are involved in extracellular and systemic functions such as systemic inflammation and extracellular matrix degradation. Ischemia reperfusion (IR) injury is responsible for numerous diseases including myocardial infarction, acute kidney injury, stroke and acute graft failure after transplant surgery. Inflammation plays a major role in the reperfusion phase of IR injury and previous research has shown that cathepsins are key mediators of the inflammation cascade as well as apoptosis. Taken together, cathepsins modulation could provide potential therapeutic approaches to attenuate IR injury. The present review summarized the current understanding of various cathepsin subtypes, their major physiologic functions, their roles in multi­organ IR injury and detailed selective cathepsin inhibitors with therapeutic potential.


Subject(s)
Cathepsins , Reperfusion Injury , Humans , Reperfusion Injury/metabolism , Apoptosis/physiology , Lysosomes/metabolism , Inflammation/metabolism
3.
JCI Insight ; 5(19)2020 09 17.
Article in English | MEDLINE | ID: mdl-32941183

ABSTRACT

We determined that renal proximal tubular (PT) NF-κB essential modulator (NEMO) plays a direct and critical role in ischemic acute kidney injury (AKI) using mice lacking renal PT NEMO and by targeted renal PT NEMO inhibition with mesoscale nanoparticle-encapsulated NEMO binding peptide (NBP MNP). We subjected renal PT NEMO-deficient mice, WT mice, and C57BL/6 mice to sham surgery or 30 minutes of renal ischemia and reperfusion (IR). C57BL/6 mice received NBP MNP or empty MNP before renal IR injury. Mice treated with NBP MNP and mice deficient in renal PT NEMO were protected against ischemic AKI, having decreased renal tubular necrosis, inflammation, and apoptosis compared with control MNP-treated or WT mice, respectively. Recombinant peptidylarginine deiminase type 4 (rPAD4) targeted kidney PT NEMO to exacerbate ischemic AKI in that exogenous rPAD4 exacerbated renal IR injury in WT mice but not in renal PT NEMO-deficient mice. Furthermore, rPAD4 upregulated proinflammatory cytokine mRNA and NF-κB activation in freshly isolated renal proximal tubules from WT mice but not from PT NEMO-deficient mice. Taken together, our studies suggest that renal PT NEMO plays a critical role in ischemic AKI by promoting renal tubular inflammation, apoptosis, and necrosis.


Subject(s)
Acute Kidney Injury/genetics , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Necrosis/genetics , Peptides/pharmacology , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis/genetics , Carrier Proteins , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Drug Compounding , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/metabolism , Necrosis/metabolism , Necrosis/pathology , Protein-Arginine Deiminase Type 4/pharmacology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
4.
FASEB J ; 34(9): 12083-12099, 2020 09.
Article in English | MEDLINE | ID: mdl-32738096

ABSTRACT

Mice deficient in intestinal epithelial TLR9 develop small intestinal Paneth cell hyperplasia and higher Paneth cell IL-17A levels. Since small intestinal Paneth cells and IL-17A play critical roles in hepatic ischemia reperfusion (IR) injury, we tested whether mice lacking intestinal TLR9 have increased hepatic IR injury. Mice lacking intestinal TLR9 had profoundly increased liver injury after hepatic IR compared to WT mice with exacerbated hepatocyte necrosis, apoptosis, neutrophil infiltration, and inflammatory cytokine generation. Moreover, we observed increased small intestinal inflammation and apoptosis after hepatic IR in intestinal TLR9 deficient mice. As a potential explanation for increased hepatic IR injury, fecal short-chain fatty acids butyrate and propionate levels were lower in intestinal TLR9 deficient mice. Suggesting a potential therapy for hepatic IR, exogenous administration of butyrate or propionate protected against hepatic IR injury in intestinal TLR9 deficient mice. Mechanistically, butyrate induced small intestinal IL-10 expression and downregulated the claudin-2 expression. Finally, IL-10 neutralization abolished the protective effects of butyrate against hepatic IR injury. Our studies show intestinal TLR9 deficiency results in exacerbated hepatic IR injury with increased small intestinal apoptosis and inflammation. Furthermore, short-chain fatty acids butyrate and propionate protect against hepatic IR injury and intestinal apoptosis/inflammation in intestinal TLR9 deficient mice.


Subject(s)
Fatty Acids/immunology , Hepatocytes/immunology , Intestine, Small/immunology , Liver Diseases/immunology , Reperfusion Injury/immunology , Toll-Like Receptor 9/deficiency , Animals , Apoptosis/genetics , Apoptosis/immunology , Fatty Acids/genetics , Hepatocytes/pathology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Intestine, Small/pathology , Liver Diseases/genetics , Liver Diseases/pathology , Mice , Mice, Knockout , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Toll-Like Receptor 9/immunology
5.
Kidney Int ; 98(1): 76-87, 2020 07.
Article in English | MEDLINE | ID: mdl-32386967

ABSTRACT

We developed an innovative therapy for ischemic acute kidney injury with discerning kidney-targeted delivery of a selective Toll-like receptor 9 (TLR9) antagonist in mice subjected to renal ischemia reperfusion injury. Our previous studies showed that mice deficient in renal proximal tubular TLR9 were protected against renal ischemia reperfusion injury demonstrating a critical role for renal proximal tubular TLR9 in generating ischemic acute kidney injury. Herein, we used 300-400 nm polymer-based mesoscale nanoparticles that localize to the renal tubules after intravenous injection. Mice were subjected to sham surgery or 30 minutes renal ischemia and reperfusion injury after receiving mesoscale nanoparticles encapsulated with a selective TLR9 antagonist (unmethylated CpG oligonucleotide ODN2088) or mesoscale nanoparticles encapsulating a negative control oligonucleotide. Mice treated with the encapsulated TLR9 antagonist either six hours before renal ischemia, at the time of reperfusion or 1.5 hours after reperfusion were protected against ischemic acute kidney injury. The ODN2088-encapsulated nanoparticles attenuated renal tubular necrosis, inflammation, decreased proinflammatory cytokine synthesis. neutrophil and macrophage infiltration and apoptosis, decreased DNA fragmentation and caspase 3/8 activation when compared to the negative control nanoparticle treated mice. Taken together, our studies further suggest that renal proximal tubular TLR9 activation exacerbates ischemic acute kidney injury by promoting renal tubular inflammation, apoptosis and necrosis after ischemia reperfusion. Thus, our studies suggest a potential promising therapy for ischemic acute kidney injury with selective kidney tubular targeting of TLR9 using mesoscale nanoparticle-based drug delivery.


Subject(s)
Acute Kidney Injury , Nanoparticles , Reperfusion Injury , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Acute Kidney Injury/prevention & control , Animals , Apoptosis , Ischemia , Kidney , Kidney Tubules, Proximal , Mice , Mice, Inbred C57BL , Reperfusion Injury/drug therapy , Reperfusion Injury/prevention & control , Toll-Like Receptor 9/genetics
6.
FASEB J ; 34(4): 5465-5482, 2020 04.
Article in English | MEDLINE | ID: mdl-32086866

ABSTRACT

We tested the hypothesis that the P2X4 purinergic receptor (P2X4) exacerbates ischemic acute kidney injury (AKI) by promoting renal tubular inflammation after ischemia and reperfusion (IR). Supporting this, P2X4-deficient (KO) mice were protected against ischemic AKI with significantly attenuated renal tubular necrosis, inflammation, and apoptosis when compared to P2X4 wild-type (WT) mice subjected to renal IR. Furthermore, WT mice treated with P2X4 allosteric agonist ivermectin had exacerbated renal IR injury whereas P2X4 WT mice treated with a selective P2X4 antagonist (5-BDBD) were protected against ischemic AKI. Mechanistically, induction of kidney NLRP3 inflammasome signaling after renal IR was significantly attenuated in P2X4 KO mice. A P2 agonist ATPγS increased NLRP3 inflammasome signaling (NLRP3 and caspase 1 induction and IL-1ß processing) in isolated renal proximal tubule cells from WT mice whereas these increases were absent in renal proximal tubules isolated from P2X4 KO mice. Moreover, 5-BDBD attenuated ATPγS induced NLRP3 inflammasome induction in renal proximal tubules from WT mice. Finally, P2X4 agonist ivermectin induced NLRP3 inflammasome and pro-inflammatory cytokines in cultured human proximal tubule cells. Taken together, our studies suggest that renal proximal tubular P2X4 activation exacerbates ischemic AKI and promotes NLRP3 inflammasome signaling.


Subject(s)
Acute Kidney Injury/pathology , Inflammasomes/metabolism , Inflammation/pathology , Kidney Tubules, Proximal/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Purinergic P2X4/physiology , Reperfusion Injury/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Animals , Apoptosis , Cytokines/metabolism , Inflammation/etiology , Inflammation/metabolism , Kidney Tubules, Proximal/immunology , Kidney Tubules, Proximal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
7.
Am J Physiol Renal Physiol ; 318(1): F260-F272, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31813250

ABSTRACT

Small intestinal Paneth cells play a critical role in acute kidney injury (AKI) and remote organ dysfunction by synthesizing and releasing IL-17A. In addition, intestine-derived norepinephrine is a major mediator of hepatic injury and systemic inflammation in sepsis. We tested the hypothesis that small intestinal Paneth cells synthesize and release norepinephrine to exacerbate ischemic AKI. After ischemic AKI, we demonstrated larger increases in portal venous norepinephrine levels compared with plasma norepinephrine in mice, consistent with an intestinal source of norepinephrine release after renal ischemia and reperfusion. We demonstrated that murine small intestinal Paneth cells express tyrosine hydroxylase mRNA and protein, a critical rate-limiting enzyme for the synthesis of norepinephrine. We also demonstrated mRNA expression for tyrosine hydroxylase in human small intestinal Paneth cells. Moreover, freshly isolated small intestinal crypts expressed significantly higher norepinephrine levels after ischemic AKI compared with sham-operated mice. Suggesting a critical role of IL-17A in Paneth cell-mediated release of norepinephrine, recombinant IL-17A induced norepinephrine release in the small intestine of mice. Furthermore, mice deficient in Paneth cells (SOX9 villin Cre mice) have reduced plasma norepinephrine levels after ischemic AKI. Finally, supporting a critical role for norepinephrine in generating ischemic AKI, treatment with the selective α-adrenergic antagonists yohimbine and phentolamine protected against murine ischemic AKI with significantly reduced renal tubular necrosis, inflammation, and apoptosis and less hepatic dysfunction. Taken together, we identify Paneth cells as a critical source of norepinephrine release that may lead to intestinal and liver injury and systemic inflammation after AKI.


Subject(s)
Acute Kidney Injury/metabolism , Ischemia/metabolism , Kidney/metabolism , Norepinephrine/metabolism , Paneth Cells/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis/physiology , Humans , Inflammation/metabolism , Inflammation/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Ischemia/pathology , Kidney/blood supply , Kidney/pathology , Mice , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
8.
Kidney Res Clin Pract ; 38(4): 427-440, 2019 Dec 31.
Article in English | MEDLINE | ID: mdl-31537053

ABSTRACT

Acute kidney injury (AKI) due to renal ischemia reperfusion (IR) is a major clinical problem without effective therapy and is a significant and frequent cause of morbidity and mortality during the perioperative period. Although the pathophysiology of ischemic AKI is not completely understood, several important mechanisms of renal IR-induced AKI have been studied. Renal ischemia and subsequent reperfusion injury initiates signaling cascades mediating renal cell necrosis, apoptosis, and inflammation, leading to AKI. Better understanding of the molecular and cellular pathophysiological mechanisms underlying ischemic AKI will provide more targeted approach to prevent and treat renal IR injury. In this review, we summarize important mechanisms of ischemic AKI, including renal cell death pathways and the contribution of endothelial cells, epithelial cells, and leukocytes to the inflammatory response during ischemic AKI. Additionally, we provide some updated potential therapeutic targets for the prevention or treatment of ischemic AKI, including Toll-like receptors, adenosine receptors, and peptidylarginine deiminase 4. Finally, we propose mechanisms of ischemic AKI-induced liver, intestine, and kidney dysfunction and systemic inflammation mainly mediated by Paneth cell degranulation as a potential explanation for the high mortality observed with AKI.

9.
Am J Physiol Renal Physiol ; 317(3): F743-F756, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31313953

ABSTRACT

Acute kidney injury (AKI) due to renal ischemia-reperfusion (I/R) is a major clinical problem without effective therapy. Ginger is one of the most widely consumed spices in the world, and 6-shogaol, a major ginger metabolite, has anti-inflammatory effects in neuronal and epithelial cells. Here, we demonstrate our novel findings that 6-shogaol treatment protected against renal I/R injury with decreased plasma creatinine, blood urea nitrogen, and kidney neutrophil gelatinase-associated lipocalin mRNA synthesis compared with vehicle-treated mice subjected to renal I/R. Additionally, 6-shogaol treatment reduced kidney inflammation (decreased proinflammatory cytokine and chemokine synthesis as well as neutrophil infiltration) and apoptosis (decreased TUNEL-positive renal tubular cells) compared with vehicle-treated mice subjected to renal I/R. In cultured human and mouse kidney proximal tubule cells, 6-shogaol significantly attenuated TNF-α-induced inflammatory cytokine and chemokine mRNA synthesis. Mechanistically, 6-shogaol significantly attenuated TNF-α-induced NF-κB activation in human renal proximal tubule cells by reducing IKKαß/IκBα phosphorylation. Furthermore, 6-shogaol induced a cytoprotective chaperone heme oxygenase (HO)-1 via p38 MAPK activation in vitro and in vivo. Consistent with these findings, pretreatment with the HO-1 inhibitor zinc protoporphyrin IX completely prevented 6-shogaol-mediated protection against ischemic AKI in mice. Taken together, our study showed that 6-shogaol protects against ischemic AKI by attenuating NF-κB activation and inducing HO-1 expression. 6-Shogaol may provide a potential therapy for ischemic AKI during the perioperative period.


Subject(s)
Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Catechols/pharmacology , Heme Oxygenase-1/metabolism , Kidney/drug effects , Membrane Proteins/metabolism , NF-kappa B/metabolism , Reperfusion Injury/prevention & control , Acute Kidney Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Animals , Apoptosis/drug effects , Cell Line , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Heme Oxygenase-1/genetics , Humans , Inflammation Mediators/metabolism , Kidney/enzymology , Kidney/pathology , Male , Membrane Proteins/genetics , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Reperfusion Injury/enzymology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Am J Physiol Renal Physiol ; 316(6): F1180-F1190, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30943066

ABSTRACT

Peptidyl arginine deiminase-4 (PAD4) catalyzes the conversion of peptidylarginine residues to peptidylcitrulline. We have previously shown that kidney ischemia-reperfusion (I/R) injury increases renal proximal tubular PAD4 expression and activity. Furthermore, kidney PAD4 plays a critical role in ischemic acute kidney injury (AKI) by promoting renal tubular inflammation, neutrophil infiltration, and NF-κB activation. However, the mechanisms of PAD4-mediated renal tubular inflammation and NF-κB activation after I/R remain unclear. Here, we show that recombinant PAD4 preferentially citrullinates recombinant IKKγ [also called NF-κB essential modulator (NEMO)] over recombinant IKKα or IKKß. Consistent with this finding, PAD4 citrullinated renal proximal tubular cell IKKγ and promoted NF-κB activation via IκBα phosphorylation in vitro. NEMO inhibition with a selective NEMO-binding peptide attenuated PAD4-mediated proinflammatory cytokine mRNA induction in HK-2 cells. Moreover, NEMO inhibition did not affect proximal tubular cell survival, proliferation, or apoptosis, unlike global NF-κB inhibition. In vivo, NEMO-binding peptide treatment protected against ischemic AKI. Finally, NEMO-binding peptide attenuated recombinant PAD4-mediated exacerbation of ischemic AKI, renal tubular inflammation, and apoptosis. Taken together, our results show that PAD4 exacerbates ischemic AKI and inflammation by promoting renal tubular NF-κB activity and inflammation via NEMO citrullination. Targeting NEMO activation may serve as a potential therapy for this devastating clinical problem.


Subject(s)
Apoptosis , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Tubules, Proximal/enzymology , Protein-Arginine Deiminase Type 4/metabolism , Reperfusion Injury/enzymology , Animals , Cell Line , Cell Proliferation , Citrullination , Disease Models, Animal , Kidney Tubules, Proximal/pathology , Male , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Neutrophil Infiltration , Phosphorylation , Reperfusion Injury/pathology , Signal Transduction
11.
JCI Insight ; 4(4)2019 02 21.
Article in English | MEDLINE | ID: mdl-30829647

ABSTRACT

Acute cardiorenal syndrome (CRS-1) is a morbid complication of acute cardiovascular disease. Heart-to-kidney signals transmitted by "cardiorenal connectors" have been postulated, but investigation into CRS-1 has been limited by technical limitations and a paucity of models. To address these limitations, we developed a translational model of CRS-1, cardiac arrest and cardiopulmonary resuscitation (CA/CPR), and now report findings from nanoscale mass spectrometry proteomic exploration of glomerular filtrate 2 hours after CA/CPR or sham procedure. Filtrate acquisition was confirmed by imaging, molecular weight and charge distribution, and exclusion of protein specific to surrounding cells. Filtration of proteins specific to the heart was detected following CA/CPR and confirmed with mass spectrometry performed using urine collections from mice with deficient tubular endocytosis. Cardiac LIM protein was a CA/CPR-specific filtrate component. Cardiac arrest induced plasma release of cardiac LIM protein in mice and critically ill human cardiac arrest survivors, and administration of recombinant cardiac LIM protein to mice altered renal function. These findings demonstrate that glomerular filtrate is accessible to nanoscale proteomics and elucidate the population of proteins filtered 2 hours after CA/CPR. The identification of cardiac-specific proteins in renal filtrate suggests a novel signaling mechanism in CRS-1. We expect these findings to advance understanding of CRS-1.


Subject(s)
Cardio-Renal Syndrome/physiopathology , Glomerular Filtration Barrier/physiopathology , Heart Arrest/complications , LIM Domain Proteins/metabolism , Reperfusion Injury/physiopathology , Acute Disease , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cardio-Renal Syndrome/etiology , Cardio-Renal Syndrome/urine , Cardiopulmonary Resuscitation , Cell Line , Disease Models, Animal , Glomerular Filtration Barrier/diagnostic imaging , Glomerular Filtration Barrier/metabolism , Heart Arrest/therapy , Humans , Intravital Microscopy , LIM Domain Proteins/urine , Male , Mass Spectrometry/methods , Mice , Podocytes , Proteomics/methods , Reperfusion Injury/etiology , Reperfusion Injury/urine
12.
Curr Opin Pharmacol ; 47: 65-74, 2019 08.
Article in English | MEDLINE | ID: mdl-30921560

ABSTRACT

Sepsis is life-threatening systemic organ dysfunction caused by a deregulated host response to an infectious insult. Currently, the treatment of sepsis is limited to the use of antibiotics, fluids, and cardiovascular/respiratory support. Despite these interventions, septic mortality remains high, with reduced life quality in survivors. For this reason, the identification of novel drug targets is a pressing task of modern pharmacology. According to a recent research, it appears that P2 purinergic receptors, which can regulate the host's response to infections, have been identified as potential targets for the treatment of sepsis. Among P2 receptors, the P2X4 receptor has recently captured the attention of the research community owing to its role in protecting against infections, inflammation, and organ injury. The present review provides an outline of the role played by P2X4 receptors in the modulation of the host's response to sepsis and the promise that targeting this receptor holds in the treatment of sepsis.


Subject(s)
Receptors, Purinergic P2X4/immunology , Sepsis/immunology , Animals , Humans , Myeloid Cells/immunology
13.
Kidney Int ; 95(4): 859-879, 2019 04.
Article in English | MEDLINE | ID: mdl-30777286

ABSTRACT

Intestinal Paneth cells play a critical role in ischemic acute kidney injury (AKI) by releasing interleukin 17A (IL-17A). Because Toll-like receptor 9 (TLR9) activation degranulates Paneth cells and necrotic tubular epithelial cells release several damage associated molecular patterns that target TLR9, we tested the hypothesis that intestinal TLR9 deficiency would protect against ischemic AKI and associated remote intestinal and hepatic dysfunction by decreasing Paneth cell degranulation. We generated mice lacking TLR9 in intestinal epithelia (TLR9fl/fl Villin Cre mice) and compared them to wild type (TLR9fl/fl) mice following right nephrectomy and left ischemia/reperfusion. To our surprise, mice lacking intestinal TLR9 had exacerbated kidney, liver, and small intestine injury after ischemia/reperfusion compared to wild type mice, characterized by increased kidney and intestinal inflammation, apoptosis, and necrosis as well as increased hepatic inflammation and apoptosis. Mice lacking intestinal TLR9 had larger Paneth cell granule size, pronounced intestinal macrophage infiltration, and higher intestinal crypt IL-17A expression. Administration of IL-17A neutralizing antibody prevented the exacerbation of ischemic AKI in mice lacking intestinal TLR9. These studies suggest that intestinal TLR9 activation protects against ischemic AKI and associated remote multi-organ dysfunction syndrome by regulating Paneth cell IL-17A synthesis.


Subject(s)
Acute Kidney Injury/immunology , Interleukin-17/metabolism , Multiple Organ Failure/immunology , Paneth Cells/pathology , Toll-Like Receptor 9/metabolism , Acute Kidney Injury/pathology , Animals , Apoptosis , Disease Models, Animal , Disease Progression , Humans , Hyperplasia/immunology , Hyperplasia/pathology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestines/immunology , Intestines/pathology , Kidney/immunology , Kidney/pathology , Liver/immunology , Liver/pathology , Macrophages/immunology , Male , Mice , Mice, Transgenic , Multiple Organ Failure/pathology , Paneth Cells/immunology , Paneth Cells/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Toll-Like Receptor 9/genetics
14.
J Immunol ; 202(2): 527-538, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30530591

ABSTRACT

G2A is a GPCR abundantly expressed in immune cells. G2A-/- mice showed higher lethality, higher plasma cytokines, and an impaired bacterial clearance in response to a murine model of sepsis (cecal ligation and puncture), which were blocked by GdCl3, an inhibitor of Kupffer cells. Anti-IL-10 Ab reversed the impaired bacterial clearance in G2A-/- mice. Indomethacin effectively blocked both the increased i.p. IL-10 levels and the impaired bacterial clearance, indicating that disturbed PG system is the proximal cause of these phenomena. Stimulation with LPS/C5a induced an increase in Escherichia coli phagocytosis and intracellular cAMP levels in G2A+/+ peritoneal macrophages but not G2A-/- cells, which showed more PGE2/nitrite release and intracellular reactive oxygen species levels. Heterologous coexpression of G2A and adenosine receptor type 2b (A2bAR) induced a synergistic increase in cAMP signaling in a ligand-independent manner, with the evidence of physical interaction of G2A with A2bAR. BAY 60-6583, a specific agonist for A2bAR, increased intracellular cAMP levels in Kupffer cells from G2A+/+ but not from G2A-/- mice. Both G2A and A2bAR were required for antiseptic action of lysophosphatidylcholine. These results show inappropriate activation of G2A-/- Kupffer cells to septic insults due to an impaired cAMP signaling possibly by lack of interaction with A2bAR.


Subject(s)
Cell Cycle Proteins/metabolism , Escherichia coli Infections/immunology , Escherichia coli/physiology , Kupffer Cells/immunology , Macrophages, Peritoneal/physiology , Receptor, Adenosine A2B/metabolism , Receptors, G-Protein-Coupled/metabolism , Sepsis/metabolism , Animals , Antibodies, Blocking , Cell Cycle Proteins/genetics , Cells, Cultured , Cyclic AMP/metabolism , Disease Models, Animal , Humans , Interleukin-10/immunology , Interleukin-10/metabolism , Macrophages, Peritoneal/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Protein Binding , Reactive Oxygen Species/metabolism , Receptor Cross-Talk , Receptor, Adenosine A2B/genetics , Receptors, G-Protein-Coupled/genetics , Sepsis/genetics , Signal Transduction
15.
J Immunol ; 201(3): 1073-1085, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29898963

ABSTRACT

The role for kidney TLR9 in ischemic acute kidney injury (AKI) remains unclear. In this study, we tested the hypothesis that renal proximal tubular TLR9 activation exacerbates ischemic AKI by promoting renal tubular epithelial apoptosis and inflammation. To test this hypothesis, we generated mice lacking TLR9 in renal proximal tubules (TLR9fl/fl PEPCK Cre mice). Contrasting previous studies in global TLR9 knockout mice, mice lacking renal proximal tubular TLR9 were protected against renal ischemia/reperfusion (IR) injury, with reduced renal tubular necrosis, inflammation (decreased proinflammatory cytokine synthesis and neutrophil infiltration), and apoptosis (decreased DNA fragmentation and caspase activation) when compared with wild-type (TLR9fl/fl) mice. Consistent with this, a selective TLR9 agonist oligonucleotide 1668 exacerbated renal IR injury in TLR9fl/fl mice but not in renal proximal tubular TLR9-null mice. Furthermore, in cultured human and mouse proximal tubule cells, TLR9-selective ligands induced NF-κB activation, proinflammatory cytokine mRNA synthesis, as well as caspase activation. We further confirm in the present study that global TLR9 deficiency had no impact on murine ischemic AKI. Taken together, our studies show that renal proximal tubular TLR9 activation exacerbates ischemic AKI by promoting renal tubular inflammation, apoptosis as well as necrosis, after IR via NF-κB and caspase activation. Our studies further suggest the complex nature of TLR9 activation, as renal tubular epithelial TLR9 promotes cell injury and death whereas TLR9 signaling in other cell types may promote cytoprotective effects.


Subject(s)
Acute Kidney Injury/metabolism , Kidney Tubules, Proximal/metabolism , Toll-Like Receptor 9/metabolism , Animals , Apoptosis/physiology , Cells, Cultured , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Necrosis/metabolism , Neutrophil Infiltration/physiology , Signal Transduction/physiology
16.
J Cardiothorac Vasc Anesth ; 32(5): 2190-2200, 2018 10.
Article in English | MEDLINE | ID: mdl-29452878

ABSTRACT

OBJECTIVE: To evaluate the prognostic utility of multiple novel urinary biomarkers of renal injury when used alone, in pair-wise combination with an early delta serum creatinine (ΔSCr) term, and combined as a broad biomarker panel for the prediction of serious adverse outcomes that may reflect AKI in patients undergoing cardiac surgery. DESIGN: Post-hoc analysis of prospective observational study. SETTING: Academic medical center. PARTICIPANTS: 603 adults undergoing cardiac surgery. INTERVENTIONS: None. MEASUREMENTS AND MAIN RESULTS: Urinary cystatin-c, kidney injury molecule-1, chemokine (C-C motif) ligand 2 and interleukin-18 were measured at baseline and <1 hour, 3 hours and 18-24 hours after separation from cardiopulmonary bypass (CPB). ΔSCr-initial was defined as the difference in SCr from baseline to first postoperative measure. The primary outcome of hospital mortality or renal replacement therapy occurred in 25 patients. Concordant elevation of any urinary biomarker measured 3 hours after CPB together with ΔSCr-initial ≥0 mg.dL-1 provided excellent early risk stratification for the primary outcome (OR ≥15.1, 95% CI 4.1-55.4). Combining four urinary biomarkers together with ΔSCr-initial and neutrophil gelatinase-associated lipocalin, previously reported from the same cohort, to provide a 6-point AKI risk score enabled early identification of patients reaching the primary outcome (ROCAUC 0.86, 95% CI 0.79-0.92) with potentially useful sensitivity and specificity at varied cut-points. CONCLUSIONS: Combining novel urinary biomarkers of renal injury with a creatinine-based metric soon after cardiac surgery provided excellent prognostic utility for serious adverse outcomes. Future studies are required to confirm these findings and determine optimal biomarker combinations for cost-effective risk stratification.


Subject(s)
Acute Kidney Injury/blood , Cardiac Surgical Procedures/adverse effects , Creatinine/blood , Risk Assessment/methods , Acute Kidney Injury/epidemiology , Acute Kidney Injury/etiology , Aged , Biomarkers/blood , Female , Follow-Up Studies , Hospital Mortality/trends , Humans , Incidence , Male , Middle Aged , Prospective Studies , Survival Rate/trends , United States/epidemiology
17.
Am J Physiol Renal Physiol ; 314(5): F809-F819, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29357426

ABSTRACT

We previously demonstrated that kidney peptidylarginine deiminase-4 (PAD4) plays a critical role in ischemic acute kidney injury (AKI) in mice by promoting renal tubular inflammation and neutrophil infiltration (Ham A, Rabadi M, Kim M, Brown KM, Ma Z, D'Agati V, Lee HT. Am J Physiol Renal Physiol 307: F1052-F1062, 2014). Although the role of PAD4 in granulocytes including neutrophils is well known, we surprisingly observed profound renal proximal tubular PAD4 induction after renal ischemia-reperfusion (I/R) injury. Here we tested the hypothesis that renal proximal tubular PAD4 rather than myeloid-cell lineage PAD4 plays a critical role in exacerbating ischemic AKI by utilizing mice lacking PAD4 in renal proximal tubules (PAD4ff PEPCK Cre mice) or in granulocytes (PAD4ff LysM Cre mice). Mice lacking renal proximal tubular PAD4 were significantly protected against ischemic AKI compared with wild-type (PAD4ff) mice. Surprisingly, mice lacking PAD4 in myeloid cells were also protected against renal I/R injury although this protection was less compared with renal proximal tubular PAD4-deficient mice. Renal proximal tubular PAD4-deficient mice had profoundly reduced renal tubular apoptosis, whereas myeloid-cell PAD4-deficient mice showed markedly reduced renal neutrophil infiltration. Taken together, our studies suggest that both renal proximal tubular PAD4 as well as myeloid-cell lineage PAD4 play a critical role in exacerbating ischemic AKI. Renal proximal tubular PAD4 appears to contribute to ischemic AKI by promoting renal tubular apoptosis, whereas myeloid-cell PAD4 is preferentially involved in promoting neutrophil infiltration to the kidney and inflammation after renal I/R.


Subject(s)
Acute Kidney Injury/enzymology , Apoptosis , Hydrolases/metabolism , Kidney Tubules, Proximal/enzymology , Neutrophil Infiltration , Neutrophils/enzymology , Reperfusion Injury/enzymology , Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Animals , Cytokines/metabolism , Hydrolases/deficiency , Hydrolases/genetics , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Mice, Knockout , Protein-Arginine Deiminase Type 4 , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Signal Transduction
18.
Am J Physiol Renal Physiol ; 314(2): F293-F305, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29021225

ABSTRACT

We previously demonstrated that renal tubular peptidylarginine deiminase-4 (PAD4) is induced after ischemia-reperfusion (IR) injury and this induction of PAD4 exacerbates ischemic acute kidney injury (AKI) by promoting renal tubular inflammation and neutrophil infiltration. However, the mechanisms of renal tubular PAD4 induction after IR remain unknown. Here, we tested the hypothesis that ATP, a proinflammatory danger-associated molecular pattern (DAMP) ligand released from necrotic cells after IR injury, induces renal tubular PAD4 and exacerbates ischemic AKI via P2 purinergic receptor activation. ATP as well as ATPγS (a nonmetabolizable ATP analog) induced PAD4 mRNA, protein, and activity in human and mouse renal proximal tubule cells. Supporting the hypothesis that ATP induces renal tubular PAD4 via P2X7 receptor activation, A804598 (a selective P2X7 receptor antagonist) blocked the ATP-mediated induction of renal tubular PAD4 whereas BzATP (a selective P2X7 receptor agonist) mimicked the effects of ATP by inducing renal tubular PAD4 expression and activity. Moreover, ATP-mediated calcium influx in renal proximal tubule cells was blocked by A804598 and was mimicked by BzATP. P2X7 activation by BzATP also induced PAD4 expression and activity in mouse kidney in vivo. Finally, supporting a critical role for PAD4 in P2X7-mediated exacerbation of renal injury, BzATP exacerbated ischemic AKI in PAD4 wild-type mice but not in PAD4-deficient mice. Taken together, our studies show that ATP induces renal tubular PAD4 via P2X7 receptor activation to exacerbate renal tubular inflammation and injury after IR.


Subject(s)
Acute Kidney Injury/chemically induced , Adenosine Triphosphate/toxicity , Hydrolases/metabolism , Kidney Tubules, Proximal/drug effects , Protein-Arginine Deiminases/metabolism , Purinergic P2X Receptor Agonists/toxicity , Receptors, Purinergic P2X7/drug effects , Reperfusion Injury/chemically induced , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Calcium Signaling/drug effects , Cell Line , Disease Models, Animal , Disease Progression , Humans , Hydrolases/deficiency , Hydrolases/genetics , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mice, Inbred C57BL , Mice, Knockout , Necrosis , Neutrophil Infiltration/drug effects , Protein Kinase C/metabolism , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases/genetics , Receptors, Purinergic P2X7/metabolism , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology
19.
Am J Physiol Renal Physiol ; 311(2): F437-49, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27335376

ABSTRACT

We previously demonstrated that renal peptidyl arginine deiminase-4 (PAD4) is induced after renal ischemia and reperfusion (I/R) injury and exacerbates acute kidney injury (AKI) by increasing the renal tubular inflammatory response. Here, we tested whether genetic ablation of PAD4 attenuates renal injury and inflammation after I/R in mice. After renal I/R, PAD4 wild-type mice develop severe AKI with large increases in plasma creatinine, neutrophil infiltration, as well as significant renal tubular necrosis, apoptosis, and proinflammatory cytokine generation. In contrast, PAD4-deficient mice are protected against ischemic AKI with reduced real tubular neutrophil infiltration, renal tubular necrosis, and apoptosis. In addition, hepatic injury and inflammation observed in PAD4 wild-type mice after renal I/R are significantly attenuated in PAD4-deficient mice. We also show that increased renal tubular PAD4 expression after renal I/R is associated with translocation of PAD4 from the nucleus to the cytosol. Consistent with PAD4 cytosolic translocation, we show increased renal tubular cytosolic peptidyl-citrullination after ischemic AKI. Mechanistically, recombinant PAD4 treatment increased nuclear translocation of NF-κB in cultured human as well as murine proximal tubule cells that is inhibited by a PAD4 inhibitor (2-chloroamidine). Taken together, our studies further support the hypothesis that renal tubular PAD4 plays a critical role in renal I/R injury by increasing the renal tubular inflammatory response and neutrophil infiltration after renal I/R perhaps by interacting with the proinflammatory transcription factor NF-κB in the cytosol and promoting its nuclear translocation.


Subject(s)
Acute Kidney Injury/genetics , Acute Kidney Injury/pathology , Hydrolases/deficiency , Hydrolases/genetics , Liver Diseases/genetics , Liver Diseases/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Animals , Cells, Cultured , Cytosol/metabolism , Humans , Hydrolases/antagonists & inhibitors , Inflammation/genetics , Inflammation/pathology , Kidney Tubules/metabolism , Kidney Tubules, Proximal/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neutrophil Infiltration , Protein Transport , Protein-Arginine Deiminase Type 4
SELECTION OF CITATIONS
SEARCH DETAIL
...